Skip to main content
Erschienen in: Respiratory Research 1/2018

Open Access 01.12.2018 | Research

Genetic analyses in a cohort of 191 pulmonary arterial hypertension patients

verfasst von: Hang Yang, Qixian Zeng, Yanyun Ma, Bingyang Liu, Qianlong Chen, Wenke Li, Changming Xiong, Zhou Zhou

Erschienen in: Respiratory Research | Ausgabe 1/2018

Abstract

Background

Pulmonary arterial hypertension (PAH) is a progressive and fatal disorder associated with high pulmonary artery pressure. Genetic testing enables early diagnosis and offers an opportunity for family screening. To identify genetic mutations and help make a precise diagnosis, we performed genetic testing in 191 probands with PAH and tried to analyze the genotype-phenotype correlation.

Methods

Initially, PAH samples (n = 119) were submitted to BMPR2 screening using Sanger sequencing. Later, we developed a PAH panel test to identify causal mutations in 13 genes related to PAH and tried to call BMPR2 copy number variations (CNVs) with the panel data. Multiplex ligation-dependent probe amplification (MLPA) was used to search for CNVs in BMPR2, ACVRL1 and ENG. Notably, EIF2AK4 gene was also involved in the panel, which allowed to distinguish pulmonary veno-occlusive disease (PVOD)/pulmonary capillary hemangiomatosis (PCH) patients from idiopathic PAH (IPAH). Characteristics of patients were compared using t test for continuous variables.

Results

Pathogenic BMPR2 mutations were detected most frequently in 32 (17.9%) IPAH and 5 (41.7%) heritable PAH (HPAH) patients by sequencing, and 12 BMPR2 CNVs called from the panel data were all successfully confirmed by MLPA analysis. In addition, homozygous or compound heterozygous EIF2AK4 mutations were identified in 6 patients, who should be corrected to a diagnosis of PVOD/PCH. Genotype-phenotype correlation analysis revealed that PAH patients with BMPR2 mutations were younger at diagnosis (27.2y vs. 31.6y, p = 0.0003) and exhibited more severe pulmonary hemodynamic impairment and a worse cardiac index compared with those without BMPR2 mutations.

Conclusions

The panel assay represented a highly valuable tool in PAH genetic testing, not only for the detection of small sequence alterations, but also for an indication of BMPR2 CNVs, which had implications for the specific samples to perform further MLPA assay. Analyses of PAH causal genes have a great help to clinical diagnosis and deep implications in disease treatment.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12931-018-0789-9) contains supplementary material, which is available to authorized users.
Abkürzungen
ACMG
American College of Medical Genetics
ACVRL1
activin A receptor like type 1
BMPR1B
bone morphogenetic protein receptor type 1B
BMPR2
bone morphogenetic protein receptor type 2
BWA
Burrow-Wheeler Aligner
CAV1
caveolin 1
CNV
copy-number variation
DLCO
diffusing capacity of the lung for carbon monoxide
EIF2AK4
eukaryotic translation initiation factor 2 alpha kinase 4
ENG
endoglin
GDF2
growth differentiation factor 2
gnomAD
Genome Aggregation Database
HHT
hereditary hemorrhagic telangiectasias
HPAH
heritable pulmonary arterial hypertension
IPAH
idiopathic pulmonary arterial hypertension
KCNA5
potassium voltage-gated channel subfamily A member 5
KCNK3
potassium two pore domain channel subfamily K member 3
MAF
Minor allele frequency
MLPA
Multiplex ligation-dependent probe amplification
NGS
Next-generation-sequencing
NOTCH3
Notch 3
PAH
Pulmonary arterial hypertension
PAWP
Pulmonary artery wedge pressure
PCH
Pulmonary capillary hemangiomatosis
PVOD
Pulmonary veno-occlusive disease
PVR
Pulmonary vessel resistance
SMAD4
SMAD family member 4
SMAD9
SMAD family member 9
TOPBP1
DNA topoisomerase II binding protein 1
WU
Wood units

Background

Pulmonary arterial hypertension (PAH) is a progressive and fatal disorder, which is diagnosed by a mean pulmonary artery pressure ≥ 25 mmHg at rest accompanied by a pulmonary artery wedge pressure (PAWP) ≤ 15 mmHg and a pulmonary vessel resistance (PVR) > 3 Wood units (WU), excluding other known causes of pulmonary hypertension, e.g., pulmonary diseases, thromboembolic diseases and left heart diseases [1]. Given the nonspecific clinical manifestations in the early stage, patients with PAH are frequently diagnosed late. Whenever the pulmonary pathologic changes become advanced and irreversible, prognosis and survival are poor and pessimistic. It has been reported that more than 20% patients have a two-year delay in the diagnosis of PAH [2], and if not treated timely, patients might progress into right heart failure [3] and the average survival duration after diagnosis is 2.8 years [4]. Idiopathic PAH (IPAH) corresponds to sporadic disease, without any familial history of PAH or known causes, which has an estimated prevalence of 5.9/1,000,000. Heritable PAH (HPAH) is diagnosed when there is a positive family history or when a pathogenic variant has been identified. For individuals with PAH, the most effective therapy to date is continuous injection of prostacyclin analogs. Other options include calcium channel blockers, oral prostanoids, phosphodiesterase type 5 inhibitors, and endothelin receptor antagonists [5]. Lung transplantation is effective, but long-term post-surgical survival is limited [6].
Since the 1990s, much has been learned about the molecular and genetic factors related to PAH. BMPR2 mutation is identified as the main genetic cause of PAH, accounting for 75%–90% of familial PAH [7, 8] and 3.5%–40% of sporadic cases [7, 9], with an autosomal dominant inheritance pattern [10]. The BMPR2 gene encodes bone morphogenetic protein receptor type II, which belongs to the transforming growth factor-β (TGF-β) superfamily and is involved in the regulation of cell growth and apoptosis. Two other genes, ACVRL1 [11] and ENG [12], which also encode receptors belonging to the TGF-β superfamily, have been more recently identified in PAH accompanying hereditary hemorrhagic telangiectasias (HHT). Similarly to BMPR2, these two genes affect vascular proliferation. With the development of genetics, more genes have been discovered to be implicated in the pathogenesis of PAH, including KCNK3 [13], CAV1 [14], SMAD9 [15], BMPR1B [16], but considerably less commonly so (1%–3%). The discovery of a causative mutation could help make an early diagnosis and provide an opportunity for family screening [17].
Pulmonary veno-occlusive disease (PVOD) is a rare disease that shares several clinical and hemodynamic similarities with IPAH but is distinct from IPAH in pathology and prognosis. An accurate diagnosis of PVOD in the early stage is crucial for suitable drug therapy and timely option for lung transplantation. Histological examination of lung biopsy is the gold standard for the diagnosis of PVOD, but it is often too invasive and unacceptable for the patient. Recent discoveries have indicated that gene mutations in eukaryotic translation initiation factor 2 alpha kinase 4 (EIF2AK4) are responsible for inherited PVOD and pulmonary capillary hemangiomatosis (PCH) [18], and the 2015 ESC/ERS Guidelines declared that identification of a biallelic EIF2AK4 mutation was sufficient to make a diagnosis of PVOD/PCH without histological confirmation [1].
To identify the causal mutations in PAH patients and differentiate PVOD patients from those with IPAH, a custom gene panel targeting PAH and related diseases was used to test 191 probands with clinically suspected IPAH and HPAH. Furthermore, the performance of calling copy number variations (CNVs) from our panel data was evaluated.

Methods

Subjects

One hundred and ninety-one PAH patients referred by the Center of Pulmonary Vascular Disease at Fuwai Hospital between 2016 and 2017 were enrolled in our study. All patients underwent a detailed clinical examination, and other known causes of pulmonary hypertension were excluded by an expert physician.

Sanger sequencing

Genomic DNA was extracted from EDTA–anticoagulated whole blood using a Wizard® Genomic Purification System A1125 (Promega, USA) kit according to the manufacturer’s recommended protocol. All 13 exons in the BMPR2 coding sequence region and a minimum of 20 base pairs of intronic DNA flanking each exon were amplified by PCR (Tiangen Biotech, Beijing, China). Briefly, the PCR program consisted of initial denaturation for 10 min at 95 °C; followed by 30 cycles of 30 s at 95 °C, 30 s at 55 °C and 30 s at 72 °C; and a final extension at 72 °C for 7 min. Then, the PCR products were purified with a TIANgel Midi Purification Kit (Tiangen Biotech, Beijing, China) and subsequently sequenced using a BigDye Terminator Cycle sequencing kit V 3.1 (ABI Biosystems) on an ABI PRISM 3730 Sequence Analyzer according to the manufacturer’s directions.

Gene panel testing

A total of 13 genes associated with heritable PAH and related diseases, such as HHT and PVOD/ PCH were involved in our panel. Referring to GeneReview, HGMD, OMIM and literatures, well established causative genes (PAH: BMPR2 (bone morphogenetic protein receptor type 2) [19], KCNK3 (potassium two pore domain channel subfamily K member 3) [13], CAV1 (caveolin 1) [14], SMAD9 (SMAD family member 9) [15], BMPR1B (bone morphogenetic protein receptor type 1B) [16]; HHT: ACVRL1 (activin A receptor like type 1) [11], ENG (endoglin) [12], GDF2 (growth differentiation factor 2) [20], SMAD4 (SMAD family member 4) [21]; PVOD/PCH: EIF2AK4 (eukaryotic translation initiation factor 2 alpha kinase 4) [18]) and another three strong candidate genes with supportive functional studies, KCNA5 (potassium voltage-gated channel subfamily A member 5) [22], NOTCH3 (notch 3) [23], and TOPBP1 (DNA topoisomerase II binding protein 1) [24], were included. A custom-designed gene panel (Roche NimbleGen, USA) was used for gene testing. The size of the panel was 87 kb, with a coverage of 98.7% of the target regions.
Library preparation was performed according to the manufacturer’s instructions (NEBNext Ultra DNA Library Prep Kit, Illumina, USA). Briefly, 1 μg of genomic DNA was sheared to 200 bp fragments, ligated with adaptors and size-selected for PCR amplification. Every 4 amplified DNA samples were pooled and captured using the customized gene panel with biotinylated oligos (SeqCap EZ Choice Library, Roche NimbleGen, USA). The enrichment libraries were sequenced by an Illumina MiSeq 2000 sequencer (Illumina, San Diego, CA) using 2 × 150 paired-end sequencing (MiSeq Reagent Kit v2, Illumina, San Diego, CA).

Bioinformatics analysis

Only high-quality reads were retrieved by filtering out low-quality reads and adaptor sequences using Trimmomatic software. The clean read sequences were aligned to the human reference genome (hg19) by Burrow-Wheeler Aligner (BWA), and PCR duplicates were marked by the Picard software. SNPs and insertions/deletions were identified using the GATK HaplotypeCaller program (http://​www.​broadinstitute.​org/​gsa/​wiki/​index.​php/​Home_​Page) and further annotated with comprehensive ANNOVAR software for their frequencies in the Genome Aggregation Database (gnomAD) for the pathogenicity and splicing-altering prediction of single nucleotide variants in the dbNSFP database, which included results from SIFT, Ployphen-2, MutationTaster, the dbscSNV database. In addition, the clinical significance of the sequences was annotated using Clinvar (http://​www.​ncbi.​nlm.​nih.​gov/​clinvar/​), OMIM (http://​omim.​org/), Uniprot (http://​www.​uniprot.​org/) and HGMD (http://​www.​hgmd.​org). Variants with cutoff values greater than 0.6 in the dbscSNV database were defined as splice-altering. Other synonymous variants that did not fulfill the above mentioned conditions were removed. Considering that most genes related to PAH were autosomal-dominant inherited, variants with a minor allele frequency (MAF) of > 0.1% in panel genes other than EIF2AK4 were excluded from further analysis as polymorphism variants, while the variants in EIF2AK4 with a MAF of > 1% were excluded because of an autosomal-recessive inherited pattern.

Variant classification

Variants were analyzed for pathogenicity according to the recommendations of the American College of Medical Genetics (ACMG). Specifically, the analysis was based on the following criteria: (i) whether they were previously reported by a functional study or family segregation study; (ii) the nature of the variant (e.g., nonsense, frameshift indel, or splicing mutations (intron ±1 or ± 2)); (iii) variant frequency in population databases; (iv) conservation of the altered residue; (v) in silico prediction (SIFT, PholyPhen2, or MutationTaster); (vi) de novo mutation; and (vii) family segregation studies. Based on this information, a variant was classified into one of the 5 following categories: benign, likely benign, unknown significance, likely pathogenic or pathogenic.

Copy-number variation calling

All CNVs were called by panelcn.MOPS [25], which was designed to detect targeted next-generation-sequencing (NGS) panel data. Sequencing quality was checked, and the mean coverage of sequencing was 98.7%. Duplications were removed by the Picard software before CNV calling. Samples with a high correlation of read counts (RCs) were selected automatically as controls from all the samples by panelcn.MOPS. Other parameters were kept default.

Multiplex ligation-dependent probe amplification (MLPA)

To detect the CNVs in BMPR2, the commercially available MLPA kit P093-C2 was used, containing probes for BMPR2, ENG and ACVRL1 (MRC-Holland, Amsterdam, The Netherlands). MLPA was performed according to the manufacturer’s protocol (www.​mlpa.​com), and the generated fragments were detected using an ABI 3500XL DX capillary electrophoresis system (Applied Biosystems). The results were analyzed with the Coffalyser software (MRC-Holland).

Statistical analysis

Unpaired t-test was used to analyze the difference of continuous variables between different genotypes by GraphPad Prism 5. The data were presented as mean ± standard deviation (SD) and p < 0.05 was considered statistically significant. All statistical tests were two-sided.

Results

A total of 191 patients were enrolled in our study and underwent genetic testing. Of the patients, 179 suffered from IPAH and 12 had HPAH. The main clinical findings are summarized in Table 1.
Table 1
Characteristics of patients with IPAH and HPAH who underwent a genetic testing
Characteristic
IPAH (n = 179)
HPAH (n = 12)
Age at diagnosis, y
30.7 ± 10.6
24.8 ± 7.2
Female sex
135 (75.4%)
7 (58.3%)
NYHA I-II
82 (45.8%)
6 (50.0%)
NYHA III
91 (50.8%)
5 (41.7%)
NYHA IV
6 (3.4%)
1 (8.3%)
Hemodynamics at diagnosis
RAP, mmHg
5.2 ± 4.2
2.8 ± 4.5
mPAP, mmHg
56.1 ± 14.7
61.6 ± 16.4
PVR, dyn*s*cm− 5
1155.6 ± 523.6
1052.8 ± 422.4
PAWP, mmHg
7.1 ± 3.5
7.1 ± 3.1
CI, L/min/m2
2.8 ± 0.9
3.1 ± 1.2
SvO2, %
67.8 ± 7.5
71.4 ± 6.3
Peak VO2, ml/min/Kg
13.1 ± 3.6
12.2 ± 2.2
6-min walk distance, m
415.6 ± 101.3
435.5 ± 100.3
NT-proBNP, pg/ml
1403.6 ± 1327.4
970.1 ± 881.8
Values are mean ± SD or n (%). NYHA, New York Heart Association functional class; RAP right atrial pressure, mPAP mean pulmonary artery pressure, PVR pulmonary vascular resistance, PAWP pulmonary artery wedge pressure, CI cardiac index, SvO2 mixed venous oxygen saturation, Peak VO2 peak oxygen consumption, NT-proBNP N-terminal pro–B-type natriuretic peptide
All samples were completed to perform high-quality genetic testing. Initially, a total of 119 PAH samples were submitted to single-gene testing of BMPR2 by Sanger sequencing. Later, we developed a PAH panel test in our lab. Then 97 samples with negative results in the BMPR2 test and 72 subsequently referred samples were then directed to a panel test involving 13 genes related to PAH. If the samples were still negative, MLPA testing was continued to search for the causative mutation (Additional file 1: Figure S1).
Sequencing of the 13 PAH genes (Additional file 1: Table S1) in the 169 probands yielded a mean depth of ~ 350× and coverage of 98.7%. Exons with low (< 20×) or no coverage were subjected to Sanger sequencing to obtain 100% coverage in the main gene, BMPR2. In addition, potential pathogenic mutations and rare variants of unknown significance (VUS) were confirmed using Sanger sequencing.
Causal mutations identified in the sequencing tests (both BMPR2 sequencing and panel sequencing), except for biallelic EIF2AK4, are described in Table 2. The table indicates that BMPR2 mutations were detected most frequently in 32 (17.9%) IPAH and 5 (41.7%) HPAH patients, while VUS are summarized in Additional file 1: Table S2. Notably, 4 HPAH families in our cohort indicated no suspected mutations either by panel sequencing or MLPA detection (Additional file 1: Figure S2), which required further genetic testing for the identification of new genes potentially related to PAH. In addition, in 6 patients clinically diagnosed with IPAH, homozygous or compound heterozygous EIF2AK4 mutations were detected (Table 3); thus, their diagnosis had to be corrected to PVOD/PCH.
Table 2
Pathogenic and likely pathogenic mutations in PAH-related genes except for EIF2AK4
Patient
Gene
Transcript
Exon/Intron
Nucleotide change
Protein change
De novo
Pathogenicity
Ref PMID
MAF
PAH1
BMPR2
NM_001204
exon5
c.551dupA
p.His184fs
NA
P
NA
NA
PAH10
BMPR2
NM_001204
exon6
c.793_796del
p.Glu265fs
NA
P
NA
NA
PAH17
BMPR2
NM_001204
exon6
c.631C > T
p.Arg211Term
NA
P
NA
NA
PAH25
ACVRL1
NM_001077401
exon6
c.994A > T
p.Lys332Term
NA
P
NA
NA
PAH27
BMPR2
NM_001204
exon3
c.338dupA
p.Tyr113_Arg114delinsTerm
NA
P
NA
NA
PAH31
BMPR2
NM_001204
exon4
c.439C > T
p.Arg147Term
NA
P
11,115,378
8.96 × 10−6
PAH34
BMPR2
NM_001204
exon6
c.705delA
p.Asn236fs
NA
P
NA
NA
PAH39
NOTCH3
NM_000435
exon11
c.1493–2- > C
 
NA
P
NA
9.78 × 10−5
PAH42
BMPR2
NM_001204
exon7
c.893delG
p.Val299Term
NA
P
NA
NA
PAH43
BMPR2
NM_001204
exon1
c.71_76del
p.24_26del
NA
LP
NA
NA
PAH46
BMPR2
NM_001204
exon6
c.631C > T
p.Arg211Term
De novo
P
11,115,378
NA
PAH47
BMPR2
NM_001204
exon12
c.2567delC
p.Thr856fs
NA
P
NA
NA
PAH51
BMPR2
NM_001204
exon10
c.1372C > T
p.Gln458Term
NA
P
21,737,554
NA
PAH67
BMPR2
NM_001204
exon7
c.944 T > C
p.Leu315P
De novo
LP
NA
NA
PAH75
BMPR2
NM_001204
exon8
c.1089delG
p.Val364fs
Inherited
P
NA
NA
PAH76
BMPR2
NM_001204
exon1
c.47G > A
p.Trp16Term
NA
P
15,687,131
NA
PAH86
ACVRL1
NM_001077401
exon9
c.1450C > T
p.Arg484Trp
De novo
P
11,484,689
NA
PAH95
ENG
NM_000118
exon3
c.247C > T
p.Gln83Term
NA
P
15,517,393
NA
PAH98
BMPR2
NM_001204
exon6
c.631C > T
p.Arg211Term
NA
P
11,015,450
NA
PAH102
BMPR2
NM_001204
exon12
c.1730 T > A
p.Leu577Term
NA
LP
NA
NA
PAH110
BMPR2
NM_001204
exon11
c.1471C > T
p.Arg491Trp
NA
LP
NA
NA
PAH117
BMPR2
NM_001204
exon2
c.178 T > C
p.Cys60Arg
NA
LP
NA
NA
PAH123
BMPR2
NM_001204
intron3
c.418 + 4A > G
 
NA
LP
NA
NA
PAH137
ACVRL1
NM_001077401
exon9
c.1450C > T
p.Arg484Trp
NA
P
11,484,689
NA
PAH138
BMPR2
NM_001204
exon9
c.1249insAT
p.Phe417fs
NA
P
NA
NA
PAH143
BMPR2
NM_001204
exon12
c.1789C > T
p.Arg597Term
NA
P
NA
NA
PAH150
BMPR2
NM_001204
exon8
c.994C > T
p.Arg332Term
NA
P
11,015,450
NA
PAH153
BMPR2
NM_001204
exon12
c.2617C > T
p.Arg873Term
NA
P
10,903,931
NA
PAH155
BMPR2
NM_001204
exon12
c.1789C > T
p.Arg597Term
NA
P
NA
NA
PAH159
BMPR2
NM_001204
exon12
c.2470C > T
p.Gln824Term
Inherited
LP
NA
NA
PAH163
BMPR2
NM_001204
exon12
c.2522 delA
p.His841fs
NA
P
18,356,561
NA
PAH166
BMPR2
NM_001204
exon12
c.2710_2831dup
p.Ala945fs
NA
P
NA
NA
PAH168
ENG
NM_000118
exon12
c.1503_1504insG
p.Gly501fs
NA
P
NA
NA
PAH176
BMPR2
NM_001204
exon6
c.631C > T
p.Arg211Term
NA
P
11,015,450
NA
PAH177
BMPR2
NM_001204
exon6
c.631C > T
p.Arg211Term
NA
P
11,015,450
NA
PAH178
BMPR2
NM_001204
exon6
c.713_714del
p.238_238del
Inherited
LP
NA
NA
PAH182
ACVRL1
NM_001077401
exon7
c.1232G > A
p.Arg411Gln
Inherited
LP
8,640,225
6.55 × 10−5
PAH183
BMPR2
NM_001204
intron6
IVS6 + 3A > C
 
NA
LP
NA
NA
PAH184
BMPR2
NM_001204
exon4
c.439C > T
p.Arg147Term
NA
LP
11,115,378
8.96 × 10−6
PAH187
BMPR2
NM_001204
exon8
c.1089delG
p.Leu363fs
NA
P
NA
NA
PAH188
ACVRL1
NM_001077401
exon7
c.1121G > A
p.Arg374Gln
NA
LP
12,700,602
4.49 × 10− 5
PAH189
ACVRL1
NM_001077401
exon3
c.372delC
p.Gly124fs
De novo
P
NA
NA
PAH192
BMPR2
NM_001204
exon12
c.2268_2269insA
p.Thr756fs
NA
P
NA
NA
PAH194
BMPR2
NM_001204
exon9
c.1243G > A
p.Glu415Lys
NA
LP
NA
NA
PAH195
BMPR2
NM_001204
exon9
c.1161delG
p.Val387fs
De novo
P
NA
NA
PAH207
BMPR2
NM_001204
exon7
c.901 T > C
p.Ser301Pro
De novo
LP
16,429,395
NA
fs frame shift, NA not available, LP likely pathogenic, P pathogenic, ref. PMID reference PubMed unique identifier, MAF minor allele frequency in the Genome Aggregation Database (gnomAD)
Table 3
Pathogenic biallelic EIF2AK4 mutations
Patient ID
Sex/Age
Initial Diagnosis
Variant (1)
Variant (2)
Final Diagnosis
PAH3
F/21
IPAH
c.2403 + 1G > A, het
c.2632-1G > A, het
PVOD
PAH48
F/39
IPAH
c.2965C > T: p.Arg989Trp, het
c.4724 T > C: p.Leu1575Pro, het
PVOD
PAH151
M/26
IPAH
c.4414_4417del: p.1472_1473del, hom
 
PVOD
PAH169
F/51
IPAH
c.168delT: p.Asn56fs, het
c.4660-1G > A, het
PVOD
PAH197
F/37
IPAH
c.594 + 1G > A, het
c.2965C > T: p.Arg989Trp, het
PVOD
PAH202
F/29
IPAH
c.986_987del: p.329_329del, hom
 
PVOD
F Female, M male, het heterozygous, hom homozygous
Because deletion/duplication contributed considerably to BMPR2 pathogenic mutations in HPAH, we tried to detect CNVs in targeted NGS panel data. With the newly developed panelcn.MOPS pipeline by Povysil et al. [25], 12 BMPR2 deletions were detected, after excluding those flagged as low quality (Table 4). To confirm the results, MLPA was performed. Almost all BMPR2 CNVs were proved to be true, except for a slight inconsistency in patient PAH52, which panelcn.MOPS suggested only exon 11 del while MLPA revealed an exon 11–12 del in BMPR2, suggesting that the method implemented by panelcn.MOPS was highly effective for BMPR2 CNV detection. Confusingly, CNVs in ENG and ACVRL1 called by panelcn.MOPS were proved to be false (Additional file 1: Table S3), probably because of a high GC content (Additional file 1: Figure S3).
Table 4
CNVs in BMPR2 by panelcn.MOPS and MLPA
Patient ID
Gene
Transcript
panelcn.MOPS
MLPA
Accordance
PAH22
BMPR2
NM_001204
Ex1 del, het
Ex1 del, het
Full
PAH36
BMPR2
NM_001204
Ex4 del, het
Ex4 del, het
Full
PAH44
BMPR2
NM_001204
The whole gene del, het
The whole gene del, het
Full
PAH52
BMPR2
NM_001204
Ex11 del, het
Ex11–12 del, het
Partly
PAH60
BMPR2
NM_001204
Ex1 del, het
Ex1 del, het
Full
PAH84
BMPR2
NM_001204
Ex6–7 del, het
Ex6–7 del, het
Full
PAH100
BMPR2
NM_001204
Ex11 del, het
Ex11 del, het
Full
PAH109
BMPR2
NM_001204
The whole gene del, het
The whole gene del, het
Full
PAH158
BMPR2
NM_001204
Ex1 del, het
Ex1 del, het
Full
PAH173
BMPR2
NM_001204
Ex2–3 del, het
Ex2–3 del, het
Full
PAH174
BMPR2
NM_001204
Ex8–9 del, het
Ex8–9 del, het
Full
PAH190
BMPR2
NM_001204
Ex1 del, het
Ex1 del, het
Full
CNVs Copy number variations, Ex Exon, het heterozygous
When trying to explore the genotype-phenotype correlation, we found that PAH patients with BMPR2 causal and rare mutations were younger at diagnosis (27.2y vs. 31.6y, p = 0.0003) and had more severe pulmonary hemodynamic impairment and a worse cardiac index (2.6 L/min/m2 vs. 3.0 L/min/m2, p = 0.0017) (Table 5) compared with those without BMPR2 mutations, in accord with previous reports [9]. In addition, patients with biallelic EIF2AK4 mutations had a lower diffusion capacity for carbon monoxide of lung (DLCO) than PAH patients (Additional file 1: Table S4).
Table 5
Genotype-phenotype correlation between PAH patients with and without a BMPR2 mutation
Characteristic
PAH with a BMPR2 mutation
PAH without a BMPR2 mutation
P value
Age at diagnosis, y
27.2 ± 9.9
31.6 ± 10.5
0.0003
Female sex
38 (67.9%)
99 (76.7%)
 
NYHA I-II
27 (48.2%)
56 (43.4%)
 
NYHA III
28 (50.0%)
67 (51.9%)
 
NYHA IV
1 (1.8%)
6 (4.7%)
 
Hemodynamics at diagnosis
RAP, mmHg
5.1 ± 4.4
5.1 ± 4.2
ns
mPAP, mmHg
60.2 ± 15.3
54.9 ± 14.7
0.0321
PVR, dyn*s*cm−5
1386.2 ± 641.4
1042.1 ± 417.0
0.0024
PAWP, mmHg
6.9 ± 3.5
7.2 ± 3.5
ns
CI, L/min/m2
2.6 ± 0.9
3.0 ± 0.9
0.0017
SvO2, %
64.8 ± 8.1
69.4 ± 6.6
0.0005
Peak VO2, ml/min/Kg
11.7 ± 2.7
13.9 ± 3.6
0.0096
6-min walk distance, m
396.8 ± 106.1
427.0 ± 97.7
ns
NT-proBNP, pg/ml
1432.1 ± 1326.2
1327.4 ± 1283.9
ns
Values are mean ± SD or n (%). NYHA New York Heart Association functional class, RAP right atrial pressure, mPAP mean pulmonary artery pressure, PVR pulmonary vascular resistance, PAWP pulmonary artery wedge pressure, CI cardiac index, SvO 2 mixed venous oxygen saturation, Peak VO 2 peak oxygen consumption, NT-proBNP N-terminal pro–B-type natriuretic peptide, ns no significance

Discussion

To the best of our knowledge, this was the largest reported cohort of Chinese PAH patients undergoing genetic testing, which greatly contributed to expand PAH genetic database in Asian population. In the present study, 31.3% (56/179) IPAH and 66.7% (8/12) HPAH patients were detected causative mutations. Targeted NGS panel assay is a highly effective clinical tool, which allows not only for the detection of small sequence alterations, but also for an indication of CNVs with specific computational algorithms. Our results showed that 37 BMPR2 mutations (37/49, 75.5%) were identified in 32 IPAH and 5 HPAH by the sequencing methods and 12 BMPR2 CNVs (12/49, 24.5%) were indicated from the panel data and finally validated by MLPA, which was distinct from Cogan’s report [8] that a higher frequency of BMPR2 mutation was detected by deletions/duplications analysis in HPAH, compared with that by sequence analysis (48% vs. 37%). Therefore, an indication of CNVs from panel data would largely save costs in testing specified samples. Our results revealed that the newly developed and free available panelcn.MOPS pipeline [25] showed high sensitivity and specificity in exploring BMPR2 CNVs, ranging from single exons to the whole gene, and all were confirmed by MLPA, except for a slight inconsistency in which panelcn.MOPS suggested a single exon 11 del while MLPA revealed an exon 11–12 del in BMPR2 in patient PAH52. Conversely, CNVs detected in the genes ENG and ACVRL1 were all proved to be false, which might be related to a high GC content in these two genes.
Notably, 86 IPAH patients and 4 HPAH families were not detected any suspected mutations in the 13 known genes involved in our panel, indicating that other genes potentially responsible for the disease might exist. In most recent years, more genes were discovered, such as KLF2 [26], SOX17 [27], AQP1 [27]. We would add these novel genes and other candidate genes related to PAH into our panel in the future, such as FOXF1 [28], RASA1 [29], TBX4 [30], ABCA3 [31], SMAD1 [15]. Furthermore, we would also dedicate in novel gene discovering through whole exome sequencing (WES) later on, especially these four HPAH families with negative results in our cohort.
PVOD, featured with extensive and diffuse constriction and/or occlusion of pulmonary veins and venules, instead of pulmonary arteriopathy, shares several clinical and hemodynamic similarities with PAH, thus often leading to a misdiagnosis of IPAH. However, PVOD patients usually have a worse prognosis, and more importantly, the administration of PAH specific therapy (vasodilators) can precipitate severe acute pulmonary edema [32]. Hence, a diagnosis of PVOD/PCH distinct from that of PAH appears extremely important and identification of biallelic EIF2AK4 mutations makes it possible. In our study, in 6 patients clinically diagnosed with IPAH, homozygous or compound heterozygous EIF2AK4 mutations were detected; thus, their diagnosis should be corrected to PVOD.
Our clinical data demonstrated that patients with biallelic EIF2AK4 mutations had a lower DLCO value, suggesting a higher probability of biallelic EIF2AK4 detection in PAH patients with DLCO values < 40% of the predicted value. However, because of our limited PVOD sample size, the cut-off value of DLCO of predicted and the corresponding detection rate required further study.
It was reported that PVOD patients carrying biallelic EIF2AK4 mutations had a worse survival compared with IPAH patients [33] and PVOD patients without EIF2AK4 mutations [34], and showed no response to PAH therapy [34]. Inconsistently, neither of our two PVOD patients with intravenous injection of prostacyclin analogues, PAH3 and PAH151, demonstrated pulmonary edema. Regretfully, none of these 6 patients underwent microscopic examination of lung tissue; therefore, we were unaware of the differences that might have existed between PVOD patients who showed a good response to PAH-targeted drugs and those who did not.
With the extensive development of molecular genetics in clinical services, increasing amounts of genetic data have been obtained. Researchers are dedicated to exploring genotype-phenotype correlations in causative genes and PAH phenotypes. A meta-analysis of 1550 PAH patients revealed that PAH patients with BMPR2 mutations presented a younger age at diagnosis with more severe disease and a younger age at death or transplantation compared with non-carriers [9]. PAH patients with ACVRL1 mutations were younger at diagnosis and had a worse prognosis than other PAH patients [35]. In accord with previous reports, our data showed that PAH patients with BMPR2 mutations were younger at diagnosis and presented more severe pulmonary hemodynamic impairment and a worse cardiac index. Probably because of the limited sample size, we did not observe any differences between the ACVRL1 mutation group and other PAH patients (data not shown). The genotype-phenotype correlation exploring might help predict prognoses based on genetic results, and lay foundation for further mechanism study and target therapy.

Conclusion

Our data further expand the PAH causative gene mutation spectrum and affirm that the panelcn.MOPS pipeline is efficient for BMPR2 CNV detection using our panel sequencing data in clinical diagnosis, which strongly suggested to perform further MLPA testing in the specific samples. This approach would effectively save costs in the routine clinical diagnosis. The target panel assay undoubtedly represents a highly valuable clinical tool and lays the foundation for further study.

Acknowledgements

We thanked all subjects who participated in this study.

Funding

This work was supported by the grant of CAMS Medical Science and Health Technology Innovation Project (2016-I2M-1-016) and Capital Health Development and Scientific Research Projects (2016–2-4036).

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.
Each individual accepting the genetic test was adequately informed and signed a consent form. The study was approved by the ethics committee of Fuwai Hospital (Approval NO.: 2017–877) and adhered to the Declaration of Helsinki.
Each individual accepting the genetic test signed a consent form and agreed to allow their anonymized samples and genetic results used for further research studies and publications.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Galie N, Humbert M, Vachiery JL, Gibbs S, Lang I, Torbicki A, Simonneau G, Peacock A, Vonk Noordegraaf A, Beghetti M, et al. 2015 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension. Rev Esp Cardiol (Engl Ed). 2016;69:177. Galie N, Humbert M, Vachiery JL, Gibbs S, Lang I, Torbicki A, Simonneau G, Peacock A, Vonk Noordegraaf A, Beghetti M, et al. 2015 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension. Rev Esp Cardiol (Engl Ed). 2016;69:177.
2.
Zurück zum Zitat Brown LM, Chen H, Halpern S, Taichman D, McGoon MD, Farber HW, Frost AE, Liou TG, Turner M, Feldkircher K, et al. Delay in recognition of pulmonary arterial hypertension: factors identified from the REVEAL registry. Chest. 2011;140:19–26.CrossRefPubMedPubMedCentral Brown LM, Chen H, Halpern S, Taichman D, McGoon MD, Farber HW, Frost AE, Liou TG, Turner M, Feldkircher K, et al. Delay in recognition of pulmonary arterial hypertension: factors identified from the REVEAL registry. Chest. 2011;140:19–26.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Galie N, Humbert M, Vachiery JL, Gibbs S, Lang I, Torbicki A, Simonneau G, Peacock A, Vonk Noordegraaf A, Beghetti M, et al. ESC/ERS guidelines for the diagnosis and treatment of pulmonary hypertension: the joint task force for the diagnosis and treatment of pulmonary hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur Heart J. 2015;2016(37):67–119. Galie N, Humbert M, Vachiery JL, Gibbs S, Lang I, Torbicki A, Simonneau G, Peacock A, Vonk Noordegraaf A, Beghetti M, et al. ESC/ERS guidelines for the diagnosis and treatment of pulmonary hypertension: the joint task force for the diagnosis and treatment of pulmonary hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur Heart J. 2015;2016(37):67–119.
4.
Zurück zum Zitat Kiely DG, Elliot CA, Sabroe I, Condliffe R. Pulmonary hypertension: diagnosis and management. BMJ. 2013;346:f2028.CrossRefPubMed Kiely DG, Elliot CA, Sabroe I, Condliffe R. Pulmonary hypertension: diagnosis and management. BMJ. 2013;346:f2028.CrossRefPubMed
5.
Zurück zum Zitat McLaughlin VV, Archer SL, Badesch DB, Barst RJ, Farber HW, Lindner JR, Mathier MA, McGoon MD, Park MH, Rosenson RS, et al. ACCF/AHA 2009 expert consensus document on pulmonary hypertension: a report of the American College of Cardiology Foundation task force on expert consensus documents and the American Heart Association: developed in collaboration with the American College of Chest Physicians, American Thoracic Society, Inc., and the Pulmonary Hypertension Association. Circulation. 2009;119:2250–94.CrossRefPubMed McLaughlin VV, Archer SL, Badesch DB, Barst RJ, Farber HW, Lindner JR, Mathier MA, McGoon MD, Park MH, Rosenson RS, et al. ACCF/AHA 2009 expert consensus document on pulmonary hypertension: a report of the American College of Cardiology Foundation task force on expert consensus documents and the American Heart Association: developed in collaboration with the American College of Chest Physicians, American Thoracic Society, Inc., and the Pulmonary Hypertension Association. Circulation. 2009;119:2250–94.CrossRefPubMed
6.
Zurück zum Zitat Benza RL, Miller DP, Barst RJ, Badesch DB, Frost AE, McGoon MD. An evaluation of long-term survival from time of diagnosis in pulmonary arterial hypertension from the REVEAL registry. Chest. 2012;142:448–56.CrossRefPubMed Benza RL, Miller DP, Barst RJ, Badesch DB, Frost AE, McGoon MD. An evaluation of long-term survival from time of diagnosis in pulmonary arterial hypertension from the REVEAL registry. Chest. 2012;142:448–56.CrossRefPubMed
7.
Zurück zum Zitat Girerd B, Montani D, Jais X, Eyries M, Yaici A, Sztrymf B, Savale L, Parent F, Coulet F, Godinas L, et al. Genetic counselling in a national referral Centre for pulmonary hypertension. Eur Respir J. 2016;47:541–52.CrossRefPubMed Girerd B, Montani D, Jais X, Eyries M, Yaici A, Sztrymf B, Savale L, Parent F, Coulet F, Godinas L, et al. Genetic counselling in a national referral Centre for pulmonary hypertension. Eur Respir J. 2016;47:541–52.CrossRefPubMed
8.
Zurück zum Zitat Cogan JD, Pauciulo MW, Batchman AP, Prince MA, Robbins IM, Hedges LK, Stanton KC, Wheeler LA, Phillips JA 3rd, Loyd JE, Nichols WC. High frequency of BMPR2 exonic deletions/duplications in familial pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;174:590–8.CrossRefPubMedPubMedCentral Cogan JD, Pauciulo MW, Batchman AP, Prince MA, Robbins IM, Hedges LK, Stanton KC, Wheeler LA, Phillips JA 3rd, Loyd JE, Nichols WC. High frequency of BMPR2 exonic deletions/duplications in familial pulmonary arterial hypertension. Am J Respir Crit Care Med. 2006;174:590–8.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Evans JD, Girerd B, Montani D, Wang XJ, Galie N, Austin ED, Elliott G, Asano K, Grunig E, Yan Y, et al. BMPR2 mutations and survival in pulmonary arterial hypertension: an individual participant data meta-analysis. Lancet Respir Med. 2016;4:129–37.CrossRefPubMedPubMedCentral Evans JD, Girerd B, Montani D, Wang XJ, Galie N, Austin ED, Elliott G, Asano K, Grunig E, Yan Y, et al. BMPR2 mutations and survival in pulmonary arterial hypertension: an individual participant data meta-analysis. Lancet Respir Med. 2016;4:129–37.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Lane KB, Machado RD, Pauciulo MW, Thomson JR, Phillips JA 3rd, Loyd JE, Nichols WC, Trembath RC. Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat Genet. 2000;26:81–4.CrossRefPubMed Lane KB, Machado RD, Pauciulo MW, Thomson JR, Phillips JA 3rd, Loyd JE, Nichols WC, Trembath RC. Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat Genet. 2000;26:81–4.CrossRefPubMed
11.
Zurück zum Zitat Harrison RE, Flanagan JA, Sankelo M, Abdalla SA, Rowell J, Machado RD, Elliott CG, Robbins IM, Olschewski H, McLaughlin V, et al. Molecular and functional analysis identifies ALK-1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic telangiectasia. J Med Genet. 2003;40:865–71.CrossRefPubMedPubMedCentral Harrison RE, Flanagan JA, Sankelo M, Abdalla SA, Rowell J, Machado RD, Elliott CG, Robbins IM, Olschewski H, McLaughlin V, et al. Molecular and functional analysis identifies ALK-1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic telangiectasia. J Med Genet. 2003;40:865–71.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Gallione CJ, Klaus DJ, Yeh EY, Stenzel TT, Xue Y, Anthony KB, McAllister KA, Baldwin MA, Berg JN, Lux A, et al. Mutation and expression analysis of the endoglin gene in hereditary hemorrhagic telangiectasia reveals null alleles. Hum Mutat. 1998;11:286–94.CrossRefPubMed Gallione CJ, Klaus DJ, Yeh EY, Stenzel TT, Xue Y, Anthony KB, McAllister KA, Baldwin MA, Berg JN, Lux A, et al. Mutation and expression analysis of the endoglin gene in hereditary hemorrhagic telangiectasia reveals null alleles. Hum Mutat. 1998;11:286–94.CrossRefPubMed
13.
Zurück zum Zitat Ma L, Roman-Campos D, Austin ED, Eyries M, Sampson KS, Soubrier F, Germain M, Tregouet DA, Borczuk A, Rosenzweig EB, et al. A novel channelopathy in pulmonary arterial hypertension. N Engl J Med. 2013;369:351–61.CrossRefPubMedPubMedCentral Ma L, Roman-Campos D, Austin ED, Eyries M, Sampson KS, Soubrier F, Germain M, Tregouet DA, Borczuk A, Rosenzweig EB, et al. A novel channelopathy in pulmonary arterial hypertension. N Engl J Med. 2013;369:351–61.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Austin ED, Ma L, LeDuc C, Berman Rosenzweig E, Borczuk A, Phillips JA 3rd, Palomero T, Sumazin P, Kim HR, Talati MH, et al. Whole exome sequencing to identify a novel gene (caveolin-1) associated with human pulmonary arterial hypertension. Circ Cardiovasc Genet. 2012;5:336–43.CrossRefPubMedPubMedCentral Austin ED, Ma L, LeDuc C, Berman Rosenzweig E, Borczuk A, Phillips JA 3rd, Palomero T, Sumazin P, Kim HR, Talati MH, et al. Whole exome sequencing to identify a novel gene (caveolin-1) associated with human pulmonary arterial hypertension. Circ Cardiovasc Genet. 2012;5:336–43.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Nasim MT, Ogo T, Ahmed M, Randall R, Chowdhury HM, Snape KM, Bradshaw TY, Southgate L, Lee GJ, Jackson I, et al. Molecular genetic characterization of SMAD signaling molecules in pulmonary arterial hypertension. Hum Mutat. 2011;32:1385–9.CrossRefPubMed Nasim MT, Ogo T, Ahmed M, Randall R, Chowdhury HM, Snape KM, Bradshaw TY, Southgate L, Lee GJ, Jackson I, et al. Molecular genetic characterization of SMAD signaling molecules in pulmonary arterial hypertension. Hum Mutat. 2011;32:1385–9.CrossRefPubMed
16.
Zurück zum Zitat Chida A, Shintani M, Nakayama T, Furutani Y, Hayama E, Inai K, Saji T, Nonoyama S, Nakanishi T. Missense mutations of the BMPR1B (ALK6) gene in childhood idiopathic pulmonary arterial hypertension. Circ J. 2012;76:1501–8.CrossRefPubMed Chida A, Shintani M, Nakayama T, Furutani Y, Hayama E, Inai K, Saji T, Nonoyama S, Nakanishi T. Missense mutations of the BMPR1B (ALK6) gene in childhood idiopathic pulmonary arterial hypertension. Circ J. 2012;76:1501–8.CrossRefPubMed
17.
Zurück zum Zitat Jones DL, Sandberg JC, Rosenthal MJ, Saunders RC, Hannig VL, Clayton EW. What patients and their relatives think about testing for BMPR2. J Genet Couns. 2008;17:452–8.CrossRefPubMedPubMedCentral Jones DL, Sandberg JC, Rosenthal MJ, Saunders RC, Hannig VL, Clayton EW. What patients and their relatives think about testing for BMPR2. J Genet Couns. 2008;17:452–8.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Eyries M, Montani D, Girerd B, Perret C, Leroy A, Lonjou C, Chelghoum N, Coulet F, Bonnet D, Dorfmuller P, et al. EIF2AK4 mutations cause pulmonary veno-occlusive disease, a recessive form of pulmonary hypertension. Nat Genet. 2014;46:65–9.CrossRefPubMed Eyries M, Montani D, Girerd B, Perret C, Leroy A, Lonjou C, Chelghoum N, Coulet F, Bonnet D, Dorfmuller P, et al. EIF2AK4 mutations cause pulmonary veno-occlusive disease, a recessive form of pulmonary hypertension. Nat Genet. 2014;46:65–9.CrossRefPubMed
19.
Zurück zum Zitat Machado RD, Pauciulo MW, Thomson JR, Lane KB, Morgan NV, Wheeler L, Phillips JA 3rd, Newman J, Williams D, Galie N, et al. BMPR2 haploinsufficiency as the inherited molecular mechanism for primary pulmonary hypertension. Am J Hum Genet. 2001;68:92–102.CrossRefPubMed Machado RD, Pauciulo MW, Thomson JR, Lane KB, Morgan NV, Wheeler L, Phillips JA 3rd, Newman J, Williams D, Galie N, et al. BMPR2 haploinsufficiency as the inherited molecular mechanism for primary pulmonary hypertension. Am J Hum Genet. 2001;68:92–102.CrossRefPubMed
20.
Zurück zum Zitat Wooderchak-Donahue WL, McDonald J, O'Fallon B, Upton PD, Li W, Roman BL, Young S, Plant P, Fulop GT, Langa C, et al. BMP9 mutations cause a vascular-anomaly syndrome with phenotypic overlap with hereditary hemorrhagic telangiectasia. Am J Hum Genet. 2013;93:530–7.CrossRefPubMedPubMedCentral Wooderchak-Donahue WL, McDonald J, O'Fallon B, Upton PD, Li W, Roman BL, Young S, Plant P, Fulop GT, Langa C, et al. BMP9 mutations cause a vascular-anomaly syndrome with phenotypic overlap with hereditary hemorrhagic telangiectasia. Am J Hum Genet. 2013;93:530–7.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Gallione CJ, Repetto GM, Legius E, Rustgi AK, Schelley SL, Tejpar S, Mitchell G, Drouin E, Westermann CJ, Marchuk DA. A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). Lancet. 2004;363:852–9.CrossRefPubMed Gallione CJ, Repetto GM, Legius E, Rustgi AK, Schelley SL, Tejpar S, Mitchell G, Drouin E, Westermann CJ, Marchuk DA. A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). Lancet. 2004;363:852–9.CrossRefPubMed
22.
Zurück zum Zitat Yuan XJ, Wang J, Juhaszova M, Gaine SP, Rubin LJ. Attenuated K+ channel gene transcription in primary pulmonary hypertension. Lancet. 1998;351:726–7.CrossRefPubMed Yuan XJ, Wang J, Juhaszova M, Gaine SP, Rubin LJ. Attenuated K+ channel gene transcription in primary pulmonary hypertension. Lancet. 1998;351:726–7.CrossRefPubMed
23.
Zurück zum Zitat Li X, Zhang X, Leathers R, Makino A, Huang C, Parsa P, Macias J, Yuan JX, Jamieson SW, Thistlethwaite PA. Notch3 signaling promotes the development of pulmonary arterial hypertension. Nat Med. 2009;15:1289–97.CrossRefPubMedPubMedCentral Li X, Zhang X, Leathers R, Makino A, Huang C, Parsa P, Macias J, Yuan JX, Jamieson SW, Thistlethwaite PA. Notch3 signaling promotes the development of pulmonary arterial hypertension. Nat Med. 2009;15:1289–97.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat de Jesus Perez VA, Yuan K, Lyuksyutova MA, Dewey F, Orcholski ME, Shuffle EM, Mathur M, Yancy L Jr, Rojas V, Li CG, et al. Whole-exome sequencing reveals TopBP1 as a novel gene in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2014;189:1260–72.CrossRefPubMedPubMedCentral de Jesus Perez VA, Yuan K, Lyuksyutova MA, Dewey F, Orcholski ME, Shuffle EM, Mathur M, Yancy L Jr, Rojas V, Li CG, et al. Whole-exome sequencing reveals TopBP1 as a novel gene in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2014;189:1260–72.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Povysil G, Tzika A, Vogt J, Haunschmid V, Messiaen L, Zschocke J, Klambauer G, Hochreiter S, Wimmer K. Panelcn.MOPS: copy-number detection in targeted NGS panel data for clinical diagnostics. Hum Mutat. 2017;38:889–97.CrossRefPubMedPubMedCentral Povysil G, Tzika A, Vogt J, Haunschmid V, Messiaen L, Zschocke J, Klambauer G, Hochreiter S, Wimmer K. Panelcn.MOPS: copy-number detection in targeted NGS panel data for clinical diagnostics. Hum Mutat. 2017;38:889–97.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Eichstaedt CA, Song J, Viales RR, Pan Z, Benjamin N, Fischer C, Hoeper MM, Ulrich S, Hinderhofer K, Grunig E. First identification of Kruppel-like factor 2 mutation in heritable pulmonary arterial hypertension. Clin Sci (Lond). 2017;131:689–98.CrossRef Eichstaedt CA, Song J, Viales RR, Pan Z, Benjamin N, Fischer C, Hoeper MM, Ulrich S, Hinderhofer K, Grunig E. First identification of Kruppel-like factor 2 mutation in heritable pulmonary arterial hypertension. Clin Sci (Lond). 2017;131:689–98.CrossRef
27.
Zurück zum Zitat Graf S, Haimel M, Bleda M, Hadinnapola C, Southgate L, Li W, Hodgson J, Liu B, Salmon RM, Southwood M, et al. Identification of rare sequence variation underlying heritable pulmonary arterial hypertension. Nat Commun. 2018;9:1416.CrossRefPubMedPubMedCentral Graf S, Haimel M, Bleda M, Hadinnapola C, Southgate L, Li W, Hodgson J, Liu B, Salmon RM, Southwood M, et al. Identification of rare sequence variation underlying heritable pulmonary arterial hypertension. Nat Commun. 2018;9:1416.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Dello Russo P, Franzoni A, Baldan F, Puppin C, De Maglio G, Pittini C, Cattarossi L, Pizzolitto S, Damante G. A 16q deletion involving FOXF1 enhancer is associated to pulmonary capillary hemangiomatosis. BMC Med Genet. 2015;16:94.CrossRefPubMedPubMedCentral Dello Russo P, Franzoni A, Baldan F, Puppin C, De Maglio G, Pittini C, Cattarossi L, Pizzolitto S, Damante G. A 16q deletion involving FOXF1 enhancer is associated to pulmonary capillary hemangiomatosis. BMC Med Genet. 2015;16:94.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Durrington HJ, Firth HV, Patient C, Belham M, Jayne D, Burrows N, Morrell NW, Chilvers ER. A novel RASA1 mutation causing capillary malformation-arteriovenous malformation (CM-AVM) presenting during pregnancy. Am J Med Genet A. 2013;161A:1690–4.CrossRefPubMed Durrington HJ, Firth HV, Patient C, Belham M, Jayne D, Burrows N, Morrell NW, Chilvers ER. A novel RASA1 mutation causing capillary malformation-arteriovenous malformation (CM-AVM) presenting during pregnancy. Am J Med Genet A. 2013;161A:1690–4.CrossRefPubMed
30.
Zurück zum Zitat Kerstjens-Frederikse WS, Bongers EM, Roofthooft MT, Leter EM, Douwes JM, Van Dijk A, Vonk-Noordegraaf A, Dijk-Bos KK, Hoefsloot LH, Hoendermis ES, et al. TBX4 mutations (small patella syndrome) are associated with childhood-onset pulmonary arterial hypertension. J Med Genet. 2013;50:500–6.CrossRefPubMedPubMedCentral Kerstjens-Frederikse WS, Bongers EM, Roofthooft MT, Leter EM, Douwes JM, Van Dijk A, Vonk-Noordegraaf A, Dijk-Bos KK, Hoefsloot LH, Hoendermis ES, et al. TBX4 mutations (small patella syndrome) are associated with childhood-onset pulmonary arterial hypertension. J Med Genet. 2013;50:500–6.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Kunig AM, Parker TA, Nogee LM, Abman SH, Kinsella JP. ABCA3 deficiency presenting as persistent pulmonary hypertension of the newborn. J Pediatr. 2007;151:322–4.CrossRefPubMed Kunig AM, Parker TA, Nogee LM, Abman SH, Kinsella JP. ABCA3 deficiency presenting as persistent pulmonary hypertension of the newborn. J Pediatr. 2007;151:322–4.CrossRefPubMed
32.
Zurück zum Zitat Ortiz-Bautista C, Hernandez-Gonzalez I, Escribano-Subias P. Pulmonary veno-occlusive disease and pulmonary capillary hemangiomatosis. Med Clin (Barc). 2017;148:265–70.CrossRef Ortiz-Bautista C, Hernandez-Gonzalez I, Escribano-Subias P. Pulmonary veno-occlusive disease and pulmonary capillary hemangiomatosis. Med Clin (Barc). 2017;148:265–70.CrossRef
33.
Zurück zum Zitat Hadinnapola C, Bleda M, Haimel M, Screaton N, Swift A, Dorfmuller P, Preston SD, Southwood M, Hernandez-Sanchez J, Martin J, et al. Phenotypic characterization of EIF2AK4 mutation carriers in a large cohort of patients diagnosed clinically with pulmonary arterial hypertension. Circulation. 2017;136:2022–33.CrossRefPubMed Hadinnapola C, Bleda M, Haimel M, Screaton N, Swift A, Dorfmuller P, Preston SD, Southwood M, Hernandez-Sanchez J, Martin J, et al. Phenotypic characterization of EIF2AK4 mutation carriers in a large cohort of patients diagnosed clinically with pulmonary arterial hypertension. Circulation. 2017;136:2022–33.CrossRefPubMed
34.
Zurück zum Zitat Montani D, Girerd B, Jais X, Levy M, Amar D, Savale L, Dorfmuller P, Seferian A, Lau EM, Eyries M, et al. Clinical phenotypes and outcomes of heritable and sporadic pulmonary veno-occlusive disease: a population-based study. Lancet Respir Med. 2017;5:125–34.CrossRefPubMed Montani D, Girerd B, Jais X, Levy M, Amar D, Savale L, Dorfmuller P, Seferian A, Lau EM, Eyries M, et al. Clinical phenotypes and outcomes of heritable and sporadic pulmonary veno-occlusive disease: a population-based study. Lancet Respir Med. 2017;5:125–34.CrossRefPubMed
35.
Zurück zum Zitat Girerd B, Montani D, Coulet F, Sztrymf B, Yaici A, Jais X, Tregouet D, Reis A, Drouin-Garraud V, Fraisse A, et al. Clinical outcomes of pulmonary arterial hypertension in patients carrying an ACVRL1 (ALK1) mutation. Am J Respir Crit Care Med. 2010;181:851–61.CrossRefPubMed Girerd B, Montani D, Coulet F, Sztrymf B, Yaici A, Jais X, Tregouet D, Reis A, Drouin-Garraud V, Fraisse A, et al. Clinical outcomes of pulmonary arterial hypertension in patients carrying an ACVRL1 (ALK1) mutation. Am J Respir Crit Care Med. 2010;181:851–61.CrossRefPubMed
Metadaten
Titel
Genetic analyses in a cohort of 191 pulmonary arterial hypertension patients
verfasst von
Hang Yang
Qixian Zeng
Yanyun Ma
Bingyang Liu
Qianlong Chen
Wenke Li
Changming Xiong
Zhou Zhou
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2018
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-018-0789-9

Weitere Artikel der Ausgabe 1/2018

Respiratory Research 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Aquatherapie bei Fibromyalgie wirksamer als Trockenübungen

03.05.2024 Fibromyalgiesyndrom Nachrichten

Bewegungs-, Dehnungs- und Entspannungsübungen im Wasser lindern die Beschwerden von Patientinnen mit Fibromyalgie besser als das Üben auf trockenem Land. Das geht aus einer spanisch-brasilianischen Vergleichsstudie hervor.

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Das Risiko für Vorhofflimmern in der Bevölkerung steigt

02.05.2024 Vorhofflimmern Nachrichten

Das Risiko, im Lauf des Lebens an Vorhofflimmern zu erkranken, ist in den vergangenen 20 Jahren gestiegen: Laut dänischen Zahlen wird es drei von zehn Personen treffen. Das hat Folgen weit über die Schlaganfallgefährdung hinaus.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.